The Effect of Genotype On Methotrexate Polyglutamate Variablity in JIA and Association With Drug Response

You might also like

Download as pdf or txt
Download as pdf or txt
You are on page 1of 10

ARTHRITIS & RHEUMATISM

Vol. 63, No. 1, January 2011, pp 276–285


DOI 10.1002/art.30080
© 2011, American College of Rheumatology

The Effect of Genotype on Methotrexate Polyglutamate


Variability in Juvenile Idiopathic Arthritis and
Association With Drug Response

Mara L. Becker,1 Roger Gaedigk,1 Leon van Haandel,2 Bradley Thomas,1 Andrew Lasky,1
Mark Hoeltzel,1 Hongying Dai,1 John Stobaugh,2 and J. Steven Leeder1

Objective. The response to and toxicity of metho- sulted in 3 distinct patterns of MTX polyglutamation.
trexate (MTX) are unpredictable in patients with juve- Cluster 1 had low red blood cell (RBC) MTXGlu
nile idiopathic arthritis (JIA). Intracellular polygluta- concentrations, cluster 2 had moderately high RBC
mation of MTX, assessed by measuring concentrations MTXGlu1 ⴙ 2 concentrations, and cluster 3 had high
of MTX polyglutamates (MTXGlu), has been demon- concentrations of MTXGlu, specifically MTXGlu3–5.
strated to be a promising predictor of drug response. SNPs in the purine and pyrimidine synthesis pathways,
Therefore, this study was aimed at investigating the as well as the adenosine pathway, were significantly
genetic predictors of MTXGlu variability and associa- associated with cluster subtype. The cluster with high
tions between MTXGlu and drug response in JIA. concentrations of MTXGlu3–5 was associated with ele-
Methods. The study was designed as a single- vated liver enzyme levels on liver function tests (LFTs),
center cross-sectional analysis of patients with JIA who and there were higher concentrations of MTXGlu3–5 in
were receiving stable doses of MTX at a tertiary care
children who reported gastrointestinal side effects and
children’s hospital. After informed consent was ob-
had abnormal findings on LFTs. No association was
tained from the 104 patients with JIA, blood was with-
noted between MTXGlu and active arthritis.
drawn during routine MTX-screening laboratory test-
Conclusion. MTXGlu remains a potentially useful
ing. Clinical data were collected by chart review.
tool for determining outcomes in patients with JIA being
Genotyping for 34 single-nucleotide polymorphisms
treated with MTX. The genetic predictors of MTXGlu
(SNPs) in 18 genes within the MTX metabolic pathway
variability may also contribute to a better understand-
was performed. An ion-pair chromatographic procedure
with mass spectrometric detection was used to measure ing of the intracellular biotransformation of MTX in
MTXGlu1–7. these patients.
Results. Analysis and genotyping of MTXGlu was
completed in the 104 patients. K-means clustering re- The response to methotrexate (MTX) in patients
with juvenile idiopathic arthritis (JIA) is variable, with
no identified predictors of response or toxicity (1–3).
Dr. Becker’s work was supported by the Katherine B. Rich- The mechanism of action of the drug is complex and
ardson Grant, the Children’s Mercy Hospital Young Investigator incompletely understood. However, over the past several
Award, the Kansas City Area Life Sciences Institute Grant, the
PhRMA Foundation Award, and the Paul Henson Immunology Re- decades, we have gained increasing knowledge regarding
search Award.
1
specific sites of MTX action within the cellular folate
Mara L. Becker, MD, MSCE, Roger Gaedigk, PhD, Bradley
Thomas, BA, Andrew Lasky, MD, Mark Hoeltzel, MD, Hongying Dai,
cycle. Due to its inhibitory effect on several target
PhD, J. Steven Leeder, PharmD, PhD: Children’s Mercy Hospitals and enzymes, we know that MTX disrupts the folate cycle,
Clinics, Kansas City, Missouri; 2Leon van Haandel, MS, John Sto- which results in decreased production of DNA precur-
baugh, PhD: University of Kansas, Lawrence.
Address correspondence to Mara L. Becker, MD, MSCE, sors, disruption of methyl-dependent reactions, and ac-
Children’s Mercy Hospitals and Clinics, Pediatrics, Section of Rheu- cumulation of adenosine, the latter of which has antiin-
matology, 2401 Gillham Road, Kansas City, MO 64108. E-mail: flammatory properties (4–6). These resultant effects of
mlbecker@cmh.edu.
Submitted for publication June 4, 2010; accepted in revised MTX have made it a useful therapy for conditions
form September 30, 2010. dependent on rapidly dividing cells for development,
276
GENOTYPE EFFECTS ON MTXGlu VARIABILITY AND DRUG RESPONSE IN JIA 277

such as cancer and psoriasis, as well as for immuno- polymerase chain reaction amplification-size discrimination
inflammatory diseases such as JIA. assays.
Collection of clinical data. Clinical response data
The polyglutamation of MTX, assessed by the included the presence or absence of active arthritis at the time
measurement of MTX polyglutamates (MTXGlu), has of the visit, the number of joints with active arthritis present at
been identified as a potentially useful biomarker of the time of the visit (defined as the active joint count), and the
treatment response in adults with rheumatoid arthritis erythrocyte sedimentation rate (ESR). Active arthritis was
(RA) (7–9). The conversion of MTX intracellularly by defined as swelling of the joints not due to bony enlargement
or, if no swelling was present, limitation of motion accompa-
folylpolyglutamate synthase to its polyglutamate forms nied by either pain on motion or tenderness that was not due
appears to be an important process for intracellular to trauma or explained by prior joint damage (15). Addition-
retention of the drug as well as for increased affinity for ally, when charts were available for review, we retrospectively
its target enzymes (4,6,10,11). We have previously re- recorded the active joint count both at 6 months and at 12
months after treatment initiation, to estimate the initial re-
ported the extensive variability in intracellular MTXGlu sponse to MTX over time. Gastrointestinal (GI) side effects
concentrations in JIA, and have observed the effect that were recorded as either 1) the presence of current GI side
route of drug administration can have on MTXGlu effects, defined as nausea, vomiting, or abdominal pain, in
concentrations (12). Due to the complexity of the folate temporal relation to the weekly MTX doses reported by the
patient or parent at the time that the blood sample was
cycle as well as the extensive variability in response to
obtained, or 2) any increased folic acid supplementation (⬎1
the drug in clinical practice, we explored potential mg/day) attributable to GI side effects in the past. In addition,
genetic factors that may affect distinct patterns of MTX- elevated liver enzyme values, defined as levels above the
Glu concentrations intracellularly, and evaluated associ- normal range on liver function tests (LFTs), were recorded
ations between the MTXGlu concentrations and drug during regular screening laboratory examinations, along with
the complete blood cell count (including the mean corpuscular
response in patients with JIA. volume).
Additional variables that were recorded as potential
confounders included age, sex, folate use, MTX dose (in
PATIENTS AND METHODS mg/kg), months receiving treatment with MTX, route of
Patient population. This study was a cross-sectional administration of MTX, use of nonsteroidal antiinflammatory
analysis of patients with JIA who were being evaluated at a drugs (NSAIDs), use of anti–tumor necrosis factor ␣ (anti-
single center. All patients diagnosed as having JIA via Inter- TNF␣) therapy (including etanercept, infliximab, and adali-
national Classification of Diseases, Ninth Revision coding mumab), and use of oral corticosteroids.
were identified, and the diagnosis was further verified through Statistical analysis. Descriptive statistics were utilized
the Edmonton 2001 International League of Associations for to characterize the variability in concentrations of each indi-
Rheumatology criteria for JIA (13). Patients between the ages vidual MTXGlu subtype (MTXGlu1–5), as described previ-
of 8 months and 21 years who were receiving a stable dose and ously (12). K-means clustering was performed as a class
discovery method for subgroup analyses. Bivariable associa-
route of MTX for a minimum of 3 months were identified for
tions were evaluated by chi-square test, Fisher’s exact test,
inclusion in the study. An additional blood sample was ob-
2-sample t-test, or Wilcoxon’s rank sum test, as appropriate.
tained at the time of a regularly scheduled blood withdrawal
Multivariate logistic regression was utilized to select significant
for laboratory testing. Parents’ written informed consent, or
risk factors and their interactions, if any, from all SNPs and
the patient’s written informed assent for those patients ages 7 covariates. Standard statistical analyses were performed using
years or older, was obtained from all participants, in accor- JMP version 8 and SAS version 9.2 (both from SAS).
dance with approval from the Children’s Mercy Hospital Multifactor dimensionality reduction (MDR) was per-
Pediatric Institutional Review Board. formed using the open-source software program mdr2.0, and
Analysis of intracellular MTX. Red blood cells (RBCs) the model’s goodness-of-fit and significance were assessed
were washed and separated as described previously (12). To using mdrpt1.0 software (16). The risk factors included 34
increase specificity and minimize interference with coeluting SNPs. Cluster outcomes and combinations were assessed sep-
substances, an ion-pair reverse-phase chromatography method arately. A subset of interactive genes was first selected using
was developed to measure the individual subtypes of MTXGlu the ReliefF filtering process. High-risk, multilocus genotype
(MTXGlu1–7) in human RBCs (14). Since MTXGlu1–5 ac- combinations, in which the ratio of cases to controls exceeded
counted for 99.6% of MTXGlutotal, subsequent analyses as- a predetermined threshold, were selected and grouped to-
sessed MTXGlu1–5. gether, which consolidated the high-dimensional risk space
Genotyping method. DNA was extracted from an into a new, 1-dimensional risk variable.
aliquot of whole blood using the Illustra blood genomicPrep The optimal one-way, two-way, and three-way MDR
Mini Spin kit (GE Healthcare). Genotyping was performed for models were determined using machine-learning techniques. A
34 single-nucleotide polymorphisms (SNPs) in 18 genes along one-locus model characterized the main effect of each SNP,
the folate/MTX metabolic pathway (Table 1), using prede- while multilocus models investigated the interactions among
signed TaqMan assays (Applied Biosystems), sequencing reac- relevant polymorphisms. A 10-fold cross-validation procedure
tions, restriction fragment length polymorphism assays, and randomly divided the whole data set into a training set (90% of
278 BECKER ET AL

Table 1. Genes and 34 associated single-nucleotide polymorphisms investigated within the methotrexate/folate metabolic pathway
Allele

Gene, rs no. Assay type* WT/var Frequency


ABCG2
rs7699188 Sequencing C/T 0.87/0.13
rs35252139 Sequencing G/A 0.87/0.13
rs35229708 Sequencing A/G 0.87/0.13
rs55930652 Sequencing C/T 0.73/027
ADORA2
rs2298383 TaqMan C/T 0.39/0.61
rs3761422 TaqMan C/T 0.67/0.33
rs2267076 TaqMan C/T 0.67/0.33
rs2236624 TaqMan C/T 0.77/0.23
ATIC
rs2372536 Sequencing C/G 0.70/0.30
rs4673990 TaqMan A/G 0.51/0.49
rs12995526 TaqMan C/T 0.54/0.46
BHMT
rs3733890 TaqMan G/A 0.67/0.33
DHFR
rs7387 RFLP A/T 0.70/0.30
19-nt del intron 1 PCR WT/var 0.64/0.36
GART
rs8971 RFLP A/G 0.81/0.19
TYMS
rs11280056 Sequencing WT/var (del) 0.68/0.32
rs34743033 PCR WT (*2)/var (*3)/var (*4) 0.485/0.510/0.005
GGH
rs1800909 TaqMan T/C 0.73/0.27
rs3758149 RFLP C/T 0.73/0.27
rs11545078 TaqMan C/T 0.92/0.08
ITPA
rs2295553 TaqMan T/C 0.48/0.52
MTHFD1
rs2236225 RFLP G/A 0.55/0.45
MTHFD2
rs56168672† RFLP WT (G)/var (del)/var (T) 0.57/0.31/0.12
16-nt del promoter† RFLP WT/var (del) 0.69/0.31
rs12196 RFLP A/G 0.66/0.34
MTHFR
rs1801133 TaqMan C/T 0.70/0.30
rs1801131 TaqMan A/C 0.67/0.33
rs2274976 RFLP G/A 0.94/0.06
MTR
rs1805087 RFLP A/G 0.81/0.19
MTRR
rs1801394 RFLP A/G 0.43/0.57
SHMT1
rs1979277 RFLP C/T 0.63/0.37
SLC01B1
rs2295553 TaqMan T/C 0.88/0.12
SLC19A1
rs1051266 RFLP A/G 0.45/0.55
SLC25A32
rs17803441 RFLP G/A 0.93/0.07

* RFLP ⫽ restriction fragment length polymorphism; PCR ⫽ polymerase chain reaction.


† Sequences for the wild-type (WT), variant (var), and 16-nucleotide deletion (del) were as follows: GTGTTGTCCGGCCTTCG-
GTGACACTAGC, GTGTTGTCCGTCCTTCGGTGACACTAGC (variant indicated in boldface), and GTGTTGdelACTAGC.

data) and a validation set (10% of data). The testing accuracy, data set. In addition, the cross-validation count was deter-
which is expressed as the percentage of true positive and true mined, which summarizes the number of times the optimal
negative actions, was obtained when the classification rule model developed from the training data set was confirmed by
developed from the training data set was applied to the testing the validation data set. Finally, the validity and significance of
GENOTYPE EFFECTS ON MTXGlu VARIABILITY AND DRUG RESPONSE IN JIA 279

Table 2. Clinical response to methotrexate within the first 12 months of treatment*


Initial 6 months 12 months
(n ⫽ 93) (n ⫽ 93) (n ⫽ 90)
Joint count, median (range)† 6 (0–63) 2 (0–48) 0 (0–42)
SC route of administration, % NA 57 68.5
Corticosteroid use, % 17.5 18.5 9.1
NSAID use, % 81.4 87 62.2
Anti-TNF␣ use, % 1 18.3 23.9
Joint count improvement, mean ⫾ SD % NA 51.6 ⫾ 59.7 72.4 ⫾ 48.0

* SC ⫽ subcutaneous; NA ⫽ not applicable; NSAID ⫽ nonsteroidal antiinflammatory drug; anti-TNF␣ ⫽


anti–tumor necrosis factor ␣.
† Number of joints with active arthritis.

the selected models were assessed by 1,000-count permutation as the American College of Rheumatology Pediatric 30
testing. (ACR Pedi 30), 50, and 70 criteria for improvement
(17). However, we were interested in how well our
RESULTS cross-sectional outcome of active or inactive disease may
represent early response to MTX. Among the 104
Demographic characteristics of the subjects. One
patients, we collected limited clinical data, including an
hundred four patients with the underlying diagnosis of
initial, 6-month, and 12-month active joint count in 93
JIA who were receiving stable doses of MTX for at least
(89.4%), 93 (89.4%), and 90 (86.5%) of the patients.
3 months were recruited between August 2007 and
August 2009. The mean ⫾ SD age in our cohort was The remaining clinical information was not accessible
117.1 ⫾ 56.5 months, and 68% were female. The major- due to the fact that the initial care of subjects was
ity of the patients (n ⫽ 97) were Caucasian (93.2%, provided at different institutions, we were unable to
including 1 of Hispanic ethnicity) and 6.7% of the retrieve the oldest records, or the 12-month data were
patients were African American. The disease duration in not yet available. The clinical characteristics of our
this cohort was a median of 33.7 months (interquartile cohort in the first 12 months of treatment with MTX,
range [IQR] 13.4–63.6 months). The median time from including the mean percentage improvement in the
diagnosis to the institution of MTX was 2.5 months active joint count at 6 months and 12 months, are
(IQR 0.89–26.9 months). The mean ⫾ SD weekly dose included in Table 2. There was no association between
of MTX was 0.51 ⫾ 0.24 mg/kg, with weekly doses coadministration of medication (including NSAIDs, glu-
ranging from 5 mg to 25 mg (mean ⫾ SD dosage 15.9 ⫾ cocorticoids, or anti-TNF␣ medications) and the per-
5.6 mg per week). The median duration of MTX therapy centage improvement in the active joint count at 6 or 12
was 18 months (IQR 9–46 months, range 3–156 months) months.
and only 44.2% of the patients reported taking supple- Subjects with active arthritis and those with inac-
mental folic acid at the time of the blood withdrawal. tive arthritis at the time that the blood sample was
Of note, 56.7% of the patients in the cohort had obtained were compared for their initial percentage of
active arthritis at the time of the examination (active improvement in active joint counts at 6 months and at 12
joint count range 1–37), and the median ESR was 12 months. Among the subjects receiving MTX, those with
mm/hour (range 1–105 mm/hour). Twenty-five percent inactive arthritis at the time of the study visit had a mean
of patients reported experiencing GI side effects in percentage improvement in the active joint count of
temporal relation to the MTX dose, and 14.4% had an 80.2% at 6 months after treatment initiation, compared
elevated value on LFTs (levels above the normal limits) with a mean percentage improvement of 31.1% in those
at the time of their concurrent screening-laboratory with active disease at the time of the study visit (P ⬍
blood withdrawal. All elevations in liver enzyme levels in 0.0001). When 12-month outcomes in relation to MTX
this cohort were ⬍2 times the upper limit of normal. treatment were compared, again those subjects with
Early response to MTX. Since the study design inactive disease at the time of our cross-sectional analy-
was cross-sectional and there were varying lengths of sis had a higher mean percentage improvement in the
MTX treatment and doses of MTX prescribed, we were active joint count at 12 months than did those who had
unable to utilize standard outcome measurements, such currently active disease (90% improvement versus 59%
280 BECKER ET AL

Table 3. Concentrations of each MTXGlu subtype according to cluster*


Cluster 1 Cluster 2 Cluster 3
(n ⫽ 50) (n ⫽ 39) (n ⫽ 15) F (P)
MTXGlu1 11.5 ⫾ 8.3 22.6 ⫾ 13.7 18.5 ⫾ 10.2 11.6 (⬍0.0001)†
MTXGlu2 8.8 ⫾ 4.7 14.4 ⫾ 4.0 15.4 ⫾ 5.2 22.6 (⬍0.0001)‡
MTXGlu3 17.6 ⫾ 9.0 45.0 ⫾ 8.7 80.4 ⫾ 14.6 254.7 (⬍0.0001)
MTXGlu4 5.7 ⫾ 5.4 16.7 ⫾ 6.7 39.5 ⫾ 8.8 160.6 (⬍0.0001)
MTXGlu5 2.0 ⫾ 2.6 4.9 ⫾ 3.4 15.3 ⫾ 8.2 60.2 (⬍0.0001)

* Clusters were determined using the K-means clustering method, with results reported as the mean ⫾ SD
nmoles/liter for each subtype of methotrexate polyglutamate (MTXGlu). Cluster associations were
assessed using the F test (with probabilities reported in parentheses), demonstrating distinct differences
between all 3 clusters or between certain groups and the remaining groups, as indicated.
† Clusters 1 and 3 versus clusters 2 and 3.
‡ Cluster 1 versus clusters 2 and 3.

improvement; P ⬍ 0.0001). When we evaluated these For example, the subjects in cluster 3, the group with the
relationships further, the mean percentage improvement highest concentrations of MTXGlu3–5, were treated with
in the active joint count at 6 months remained signifi- MTX exclusively by the subcutaneous route and re-
cantly associated with the presence of inactive disease at ceived the largest doses of MTX (mean ⫾ SD dosage
the study visit, even after controlling for MTX dose, 0.69 ⫾ 0.25 mg/kg/week) for the shortest period of time
duration of MTX, route of MTX, NSAID use, anti- (median 11 months, IQR 6–20 months). Alternatively,
TNF␣ use, initial active joint count, and percentage patients in cluster 1, who had relatively low concentra-
improvement in active joint count at 12 months (ad- tions of all MTXGlu subtypes, had the longest length of
justed P ⫽ 0.0006, R2 ⫽ 30.2). MTX treatment, for a median of 27 months (IQR
Distribution of MTXGlu subtypes. A full descrip- 10.5–55.3 months), which was significantly different
tion of MTXGlu subtype variability in JIA is discussed from that in cluster 2 (P ⫽ 0.006) and cluster 3 (P ⫽
in detail elsewhere (12). To investigate the polyglutama- 0.003).
tion patterns specific to each MTXGlu species, K-means Moreover, in comparison with that in cluster 2
clustering of the raw concentrations of each MTXGlu and cluster 3, the dose of MTX administered was
subtype was performed, and the results revealed 3 significantly lower for the subjects in cluster 1, who
distinct clusters. Cluster 1 (n ⫽ 50) had relatively low received a mean ⫾ SD dosage of 0.43 ⫾ 0.22 mg/kg/
concentrations of all subtypes (MTXGlu1–5) (mean ⫾ week (P ⫽ 0.021 versus cluster 2 and P ⫽ 0.0003 versus
SD concentration of MTXGlutotal 45.6 ⫾ 19.2 nmoles/ cluster 3); there was no statistically significant difference
liter), cluster 2 (n ⫽ 39) had moderate concentrations of in dose between cluster 2 and cluster 3 (P ⫽ 0.052).
intracellular MTXGlu1–5 (mean ⫾ SD concentration of There was no association between the current use of
MTXGlutotal 103.6 ⫾ 20.9 nmoles/liter) but relatively folic acid supplements, NSAIDs, corticosteroids, or bio-
high concentrations of native MTXGlu1, and cluster 3 logic agents and cluster assignment. Thus, the observed
(n ⫽ 15) had relatively high concentrations of pattern of MTX polyglutamation appears to be influ-
MTXGlu1–5 (mean ⫾ SD concentration of MTXGlutotal enced by dose, route of administration, and duration of
169.1 ⫾ 29.6 nmoles/liter) but significantly higher con- MTX treatment.
centrations of MTXGlu3–5 (for comparison of each To determine the contribution of genetic varia-
subtype concentration between clusters, P ⬍ 0.0001) tion to the observed patterns of MTX polyglutamation,
(Table 3). selected SNPs in genes involved in cellular folate ho-
Variables associated with MTXGlu clusters. meostasis and MTX metabolism and utilization were
Clinical variables were assessed for associations with the evaluated for associations with MTXGlu clusters. All
above-described K-means clusters. Multivariable logistic SNPs included in the analyses (34 total) were in Hardy-
regression analysis controlling for clinical variables that Weinberg equilibrium. Initially, a simple bivariable chi-
were observed to be significant (P ⬍ 0.1) in bivariable square analysis was performed to assess associations
analyses revealed that the route of administration, dose, between individual SNPs and the K-means clusters. This
and duration of MTX remained associated (R2 ⫽ 0.24) analysis identified only a marginal association with 1
with cluster distribution (P ⬍ 0.0001, P ⫽ 0.034, and P ⫽ SNP, a TYMS variant (6-nucleotide deletion at the
0.0007 for cluster 1, cluster 2, and cluster 3, respectively). 3⬘-untranslated region, rs11280056; P ⫽ 0.042), although
GENOTYPE EFFECTS ON MTXGlu VARIABILITY AND DRUG RESPONSE IN JIA 281

Figure 1. A, Distribution of methotrexate polyglutamate subtypes 1–5 (MTXGlu1–5) clustering in 2 loci,


ADORA2a (rs3761422) and ATIC (rs4673990). Values over columns are the number of alleles for ATIC, and
values beside rows are the number of alleles for ADORA2a. In each cell, open bars (and values over the bars)
represent the number of subjects in cluster 1 (low concentration of red blood cell [RBC] MTXGlu1–5), while solid
bars represent the number of subjects in cluster 3 (high concentration of RBC MTXGlu1–5). Dark gray–shaded
cells indicate the genotype combinations in which the likelihood of being in cluster 1 is relatively low, while light
gray–shaded cells indicate the genotype combinations in which the likelihood of being in cluster 3 is relatively low.
For this 2-loci model, the genetic prediction has a balanced accuracy of 80.3%, sensitivity of 74.0%, specificity of
86.7%, and precision of 99.5%. B, Radical diagram of gene–gene interactions in 3-locus multifactor dimension-
ality reduction models. The contribution of each gene (gene effects) was measured by entropy-based information
gain. Values for individual gene effects are shown in the rectangles for each single-nucleotide polymorphism
(SNP), while values for joint effects are shown in the lines connecting 2 corresponding SNPs.

this association was not significant after correction for ysis, the TYMS 6-nucleotide deletion (rs11280056) re-
multiple comparisons. mained associated with cluster outcome, whereas ATIC
To test the hypothesis that the variability in (rs4673990) and ADORA2a (rs3761422) did not show
patterns of MTX polyglutamation was a function of
epistasis, or gene–gene interactions, between folate
pathway genes, the MDR method was applied to the
Table 4. Multifactor dimensionality reduction analysis evaluating
clusters with the lowest and highest MTXGlu concen- gene–gene interactions that could differentiate cluster 1 from cluster
trations (extreme clusters 1 and 3, respectively). The 3*
most pronounced combination of SNPs differentiating Sensitivity measure Value
subjects in cluster 1 from those in cluster 3 was ATIC
(SNP rs4673990) and ADORA2a (SNP rs3761422) (Fig- Accuracy, %
Training data set 80.3
ures 1A and B). This two-way model presented the best Testing data set 80.3
overall performance, with a testing accuracy of 80.3% (P CVC 10/10
⫽ 0.006) and a cross-validation consistency of 10/10 (P ⫽ OR (95% CI) 18.5 (3.67–93.23)
P
0.0550) (Table 4). Subjects who carried this SNP com- Testing data accuracy 0.0060
bination were 18.5 times more likely to be in cluster 1 as CVC 0.0550
compared with cluster 3 (odds ratio 18.5, 95% confi- OR ⬍0.0001
dence interval 3.7–93.2; P ⬍ 0.0001). Sensitivity was * Cluster 1 was defined as subjects having low methotrexate polyglu-
74%, specificity was 86.7%, and overall accuracy was tamate (MTXGlu) concentrations of red blood cell (RBC)
MTXGlu1–5, while cluster 3 was defined as those having high concen-
80.3%. trations of RBC MTXGlu1–5, specifically, high MTXGlu3–5. The most
A multivariate logistic regression model was built pronounced combination of single-nucleotide polymorphisms differ-
with inclusion of the 3 SNPs (genotypes) that were entiating subjects in cluster 1 from those in cluster 3 was in the model
evaluating ATIC (SNP rs4673990, A⬎G) and ADORA2a (SNP
observed to be associated with cluster outcome in the rs3761422). CVC ⫽ cross-validation count; OR ⫽ odds ratio; 95%
chi-square and MDR analyses. In this multivariate anal- CI ⫽ 95% confidence interval.
282 BECKER ET AL

any statistically significant associations with cluster out- variability or, better yet, to predict the clinical response
come (P ⫽ 0.27 and P ⫽ 0.70, respectively). When have led to the measurement of intracellular MTXGlu
clinical variables that were significantly associated with concentrations as a biomarker of drug effect. Intracel-
cluster outcome were added to the model, we found that lular MTXGlu concentrations are more stable than
route of administration (P ⬍ 0.0001), dose (P ⫽ 0.043), serum levels of MTX (24), and therefore are more likely
and duration of MTX treatment (P ⫽ 0.001), in addition to be associated with treatment outcome. Long-chain
to the presence of the TYMS SNP (P ⫽ 0.021), were MTXGlu concentrations have been associated with im-
associated with cluster (R2 ⫽ 0.31). proved response in adults with RA (7,8,25), but recently,
MTXGlu polyglutamation patterns (clusters) there have been inconsistencies in the published data
and clinical outcomes. There was no association be- (26). In children, we have reported a wide variability in
tween cluster assignment and the presence of joints with intracellular MTXGlu concentrations in those with JIA,
active arthritis at the study visit, the percentage change with a strong association between route of administra-
in active joint count at 6 or 12 months, or the occurrence tion and MTXGlu chain length (12). To explore factors
of GI side effects. There were, however, differences contributing to this variability and the consequences for
between groups in the risk of an elevated liver enzyme clinical response, we herein evaluated the clinical and
value on LFTs. Cluster 1 (having the lowest MTXGlu genetic predictors of MTXGlu patterns, as well as the
concentrations) had the lowest risk of having elevated associations between MTXGlu and outcomes in patients
levels on LFTs at the time of the visit. Abnormalities on with JIA.
LFTs were observed in 4% of subjects in cluster 1 as The K-means cluster methods applied in this
compared with 23% in cluster 2 and 27% in cluster 3 analysis revealed 3 distinct clusters of MTXGlu, and
(each P ⫽ 0.014 versus cluster 1). dose, duration of MTX use, and route of administration
Further analyses of long-chain MTXGlu3–5 were all contributed to the polyglutamation pattern. One
completed. Long-chain polyglutamate concentrations, explanation for this observation may be that the clusters
corrected for the dose of MTX administered, were are populated with patients with varying levels of disease
higher in subjects who had elevated liver enzyme levels severity. For example, subjects in cluster 3 were receiv-
on LFTs at the time of their visit (mean ⫾ SD concen- ing the highest doses of MTX, for the shortest period of
tration of MTXGlu3–5 173.0 ⫾ 162.9 nmoles/liter versus time administered via the subcutaneous route, which
111.8 ⫾ 85.5 nmoles/liter in those with no abnormal may be explained if this cluster is populated with the
findings; P ⫽ 0.03) and in subjects who reported GI side most severely affected patients. There were, however, no
effects (mean ⫾ SD concentration of MTXGlu3–5 differences in the number of joints with active arthritis at
159.2 ⫾ 134.4 nmoles/liter versus 107.7 ⫾ 85.2 nmoles/ presentation (P ⫽ 0.99), at 6 months (P ⫽ 0.22), or at 12
liter in those without GI side effects; P ⫽ 0.013). No months (P ⫽ 0.43) between groups.
associations between route of administration and ele- Additionally, there were no significant differ-
vated LFT values (P ⫽ 0.14) or GI side effects (P ⫽ ences in JIA subtype, age, or the use of folic acid
0.15) were noted. In addition, there were no associations supplements between groups. However, less improve-
between dose of MTX received and elevated levels on ment in the active joint count at 6 months in cluster 3
LFTs (P ⫽ 0.35) or GI side effects (P ⫽ 0.11). (mean ⫾ SD 17 ⫾ 74% improvement) was noted
In assessing the outcome of active arthritis, there compared with that in cluster 2 (56 ⫾ 50.4% improve-
was no association between individual MTXGlu sub- ment) and cluster 1 (59.5 ⫾ 60.8% improvement) (P ⫽
types, or specifically of long-chain polyglutamates, with 0.049), and subjects in cluster 3 had a higher likelihood
favorable outcome, namely, lack of active arthritis. Ad- of having initially taken prednisone and having been
ditionally, there was no association between route and prescribed a biologic agent by 6 months, in comparison
dose of MTX with the presence of active arthritis at the with the other groups, suggesting a slower response to
time that the study sample was obtained. therapy. However, the percentage improvement in the
active joint count at 12 months was not significantly
different between groups (P ⫽ 0.97), suggesting that,
DISCUSSION
although cluster 3 may not have had as rapid an im-
Although MTX is widely administered by adult provement as the other groups, by 12 months that
and pediatric rheumatologists to treat inflammatory difference appears to have been less significant.
arthritis, there is extensive variability in response to the Our observations showing that the cluster com-
drug (2,3,18–23). Attempts to better understand this prising subjects treated with MTX for the longest period
GENOTYPE EFFECTS ON MTXGlu VARIABILITY AND DRUG RESPONSE IN JIA 283

of time had the lowest concentrations of MTXGlu, while products, which, through feedback mechanisms, may be
the cluster comprising subjects treated with MTX for the perceived as a more profound folate-depleted state.
shortest period of time had the highest concentrations of However, we cannot fully answer these questions with-
MTXGlu are the opposite of what one might expect out a better understanding of the intracellular concen-
when considering the effect of length of treatment on trations of individual folate redox states and folate
achieving steady state for the longer-chain MTXGlu polyglutamates. Therefore, in light of the knowledge
(11). We may have captured a previously underrecog- that these genes could play important roles in the
nized influence of noncompliance in the cohort of multiple processes linked to the folate pathway, we
patients with longstanding JIA; however, these patients believe that the relevance of the genes will further
were also receiving lower weekly MTX doses than were increase with our increasing knowledge about the mech-
the patients in the other 2 clusters (mean ⫾ SD 0.43 ⫾ anisms of action of MTX. In support of this notion,
0.22 mg/kg/week compared with 0.55 ⫾ 0.24 mg/kg/week recent findings have also suggested that SNPs in the
and 0.69 ⫾ 0.25 mg/kg/week in cluster 2 and cluster 3, purine synthesis pathway may be linked to MTX re-
respectively). Small numbers in cluster 3 (n ⫽ 15) may sponse in children with JIA (27) as well as those with
also have resulted in a lack of statistical power, which leukemia (28). Moreover, the results in the present study
may explain the lack of difference seen between groups may also suggest that these SNPs could play a role in
in some of the above-mentioned clinical variables. Prac- MTX biotransformation and response in JIA.
titioner style or preference for MTX dose and route of Variability in MTXGlu concentrations, however,
administration, as well as a more recent focus on early has little value if there are no associations between
and aggressive treatment, may also potentially explain MTXGlu concentrations and clinical outcome. In con-
the differences seen between clusters. These hypotheses trast to the observations reported by Dervieux and
can only be tested more clearly with a larger prospective, colleagues (7–9), and in agreement with those of
controlled study. Dolezalova and colleagues (29), we found no association
In an attempt to better elucidate the genetic between intracellular MTXGlu concentrations and the
contribution to variation and patterns in intracellular presence of active arthritis. We did, however, see a trend
MTXGlu, we utilized MDR to investigate the role of toward higher concentrations of short-chain polygluta-
gene–gene interactions (epistasis). SNPs within the pu- mates, MTXGlu1 ⫹ 2, in subjects with inactive arthritis,
rine (ATIC) and adenosine (ADORA2a) pathways were which, interestingly, is in contrast to the findings re-
found to differentiate clusters with high concentrations ported in the literature in studies of adult patients (7–9).
(specifically, MTXGlu3–5) from those with low overall However, within this cohort of patients with JIA,
concentrations. Additionally, with the use of more tra- we did find associations between MTXGlu and drug
ditional statistical methods, we identified a SNP in toxicity. The group with the lowest intracellular MTX-
TYMS that was associated with cluster outcome as well. Glu concentrations had the lowest risk of liver enzyme
We do not believe that these genes have a direct effect elevation, and the group with the highest concentration
on MTXGlu concentrations or patterns of polyglutama- of long-chain MTXGlu3–5 had the highest risk of liver
tion. Rather, we hypothesize that the observed patterns enzyme elevation. We have previously noted a higher
of polyglutamation reflect the underlying cellular folate risk of MTX toxicity in the liver in subjects with JIA who
status. The folate pathway is responsible for purine received higher doses of MTX (3); therefore, it initially
synthesis, pyrimidine synthesis, adenosine metabolism, may not be surprising that this association was found,
and homocysteine remethylation. In the presence of since cluster 3 was also the group who were receiving the
MTX and the resulting folate depletion that is the highest doses of MTX. However, even after correcting
intended therapeutic goal, the cell must allocate dwin- MTXGlu concentrations for the dose of MTX, higher
dling folate resources to specific, as yet unknown sites, concentrations of long-chain polyglutamates were seen
and therefore a better understanding of the variation in subjects with liver enzyme elevation and reported GI
(SNPs) in checkpoint genes may help to determine how side effects. These children may have inherent differ-
a pharmacologically folate-depleted cell allocates its ences in how they intracellularly transform MTX, which
resources and slows down some processes to accommo- may put them at risk for adverse effects from the drug.
date for other needs. SNP combinations that may be used to predict
Allelic variation in the genes detected, TYMS, cluster subtype may also highlight patients who will
ATIC, and ADORA2a, may result in more profound respond to MTX differently; for example, if SNPs within
inhibition of enzyme activity with reduced formation of ATIC and the adenosine receptor ADORA2a will de-
284 BECKER ET AL

crease the ability of the cell to adequately produce or limited, since it is necessary to await the eventual
respond to adenosine, these patients may have an atten- response before effectiveness or toxicity can be deter-
uated antiinflammatory response to the drug, and thus mined. We continue to search for better and more
they would have a higher likelihood of requiring higher reliable markers that could be used to predict who will
doses of MTX to control their clinical disease. Not one have a favorable response or poor response to the drug
single factor can explain the MTXGlu variability and early in the course of therapy. With the prospective
associations with outcomes, and we therefore need to studies that are being developed, we hope to gain a
determine the intracellular folate concentrations and better understanding of an individual’s baseline risk and
polyglutamate concentrations to complete the picture. ability to adapt to folate disturbances with MTX ther-
Nevertheless, this is the first study to describe an asso- apy.
ciation between MTXGlu concentrations and adverse
outcomes in relation to MTX treatment in patients with
JIA. ACKNOWLEDGMENTS
We acknowledge that our study had some limita- The authors would like to gratefully acknowledge
tions. It was designed as a cross-sectional analysis of Jaylene Weigel and Chelsey Palmer for their assistance with
patients with JIA at a single center, and thus did not patient recruitment and sample preparation, as well as Todd
allow us to dynamically follow up trends or effectively D. Williams and the University of Kansas Mass Spectrometry
facility for their guidance and collaboration in this project.
measure the response to MTX over time. The purpose
of conducting a cross-sectional and retrospective analy-
sis initially was to help us better frame the hypotheses AUTHOR CONTRIBUTIONS
and design the prospective studies to test these hypoth- All authors were involved in drafting the article or revising it
eses, which are currently in development. Our measures critically for important intellectual content, and all authors approved
the final version to be published. Dr. Becker had full access to all of the
of clinical response, including the presence and number data in the study and takes responsibility for the integrity of the data
of joints with active disease and the ESR, are not as and the accuracy of the data analysis.
rigorous as outcome measurements such as the ACR Study conception and design. Becker, Stobaugh, Leeder.
Acquisition of data. Becker, Lasky, Hoeltzel.
Pedi 30 or the Disease Activity Score. Since inactive Analysis and interpretation of data. Becker, Gaedigk, van Haandel,
disease or remission related to medications is the de- Thomas, Dai, Stobaugh, Leeder.
sired outcome of therapy in JIA, and since we were
limited by the inherent design of this pilot study, we REFERENCES
chose to evaluate the presence or absence of disease
activity, which, some may argue, is more clinically rele- 1. Cassidy JT, Petty RE, Laxer RM, Lindsley CB, editors. Textbook
of pediatric rheumatology. 5th ed. Philadelphia: Elsevier Saun-
vant. ders; 2005.
The improvement in active joint counts at 6 and 2. Ortiz-Alvarez O, Morishita K, Avery G, Green J, Petty RE,
12 months after therapy initiation helped to strengthen Tucker LB, et al. Guidelines for blood test monitoring of metho-
trexate toxicity in juvenile idiopathic arthritis. J Rheumatol 2004;
our speculations that patients who were categorized as 31:2501–6.
responders and showed a lack of active arthritis at the 3. Becker ML, Rose CD, Cron RQ, Sherry DD, Bilker WB, Laut-
time that MTXGlu was measured had a clinical course enbach E. Effectiveness and toxicity of methotrexate in juvenile
idiopathic arthritis: comparison of two initial dosing regimens.
that was more responsive to MTX. The data presented J Rheumatol 2010;37:870–5.
herein suggest that those without active arthritis at the 4. Chabner BA, Allegra CJ, Curt GA, Clendeninn NJ, Baram J,
time that the study sample was obtained had a superior Koizumi S, et al. Polyglutamation of methotrexate: is methotrexate
a prodrug? J Clin Invest 1985;76:907–12.
early response to MTX, and therefore improvement at 6 5. Cronstein BN, Naime D, Ostad E. The antiinflammatory mecha-
months may be an important factor in determining nism of methotrexate: increased adenosine release at inflamed
overall response. Although, through a cross-sectional sites diminishes leukocyte accumulation in an in vivo model of
inflammation. J Clin Invest 1993;92:2675–82.
design, we were unable to control for variables such as 6. Allegra CJ, Chabner BA, Drake JC, Lutz R, Rodbard D, Jolivet J.
MTX dose and route of administration, this relatively Enhanced inhibition of thymidylate synthase by methotrexate
large cohort of patients with JIA has provided us with polyglutamates. J Biol Chem 1985;260:9720–6.
7. Dervieux T, Furst D, Lein DO, Capps R, Smith K, Walsh M, et al.
the preliminary data and methods to develop more Polyglutamation of methotrexate with common polymorphisms in
rigorous, well-controlled prospective studies. These data reduced folate carrier, aminoimidazole carboxamide ribonucleo-
show a relationship between MTXGlu and outcomes in tide transformylase, and thymidylate synthase are associated with
methotrexate effects in rheumatoid arthritis. Arthritis Rheum
JIA, which may fuel further studies in this area. 2004;50:2766–74.
The dosing strategy for MTX in JIA remains 8. Dervieux T, Furst D, Lein DO, Capps R, Smith K, Caldwell J, et
GENOTYPE EFFECTS ON MTXGlu VARIABILITY AND DRUG RESPONSE IN JIA 285

al. Pharmacogenetic and metabolite measurements are associated rheumatoid arthritis that has been unresponsive to conventional
with clinical status in patients with rheumatoid arthritis treated doses of methotrexate: a randomized, controlled trial. Arthritis
with methotrexate: results of a multicentred cross sectional obser- Rheum 2004;50:364–71.
vational study. Ann Rheum Dis 2005;64:1180–5. 20. Cohen S, Cannon GW, Schiff M, Weaver A, Fox R, Olsen N, et al.
9. Dervieux T, Greenstein N, Kremer J. Pharmacogenomic and Two-year, blinded, randomized, controlled trial of treatment of ac-
metabolic biomarkers in the folate pathway and their association tive rheumatoid arthritis with leflunomide compared with metho-
with methotrexate effects during dosage escalation in rheumatoid trexate. Arthritis Rheum 2001;44:1984–92.
arthritis. Arthritis Rheum 2006;54:3095–103. 21. Wallace CA, Sherry DD. Preliminary report of higher dose
10. Schroder H, Fogh K. Methotrexate and its polyglutamate deriva- methotrexate treatment in juvenile rheumatoid arthritis. J Rheu-
tives in erythrocytes during and after weekly low-dose oral meth- matol 1992;19:1604–7.
otrexate therapy of children with acute lymphoblastic leukemia. 22. Alsufyani K, Ortiz-Alvarez O, Cabral DA, Tucker LB, Petty RE,
Cancer Chemother Pharmacol 1988;21:145–9. Malleson PN. The role of subcutaneous administration of metho-
11. Dalrymple JM, Stamp LK, O’Donnell JL, Chapman PT, Zhang M, trexate in children with juvenile idiopathic arthritis who have failed
Barclay ML. Pharmacokinetics of oral methotrexate in patients oral methotrexate. J Rheumatol 2004;31:179–82.
with rheumatoid arthritis. Arthritis Rheum 2008;58:3299–308. 23. Reiff A, Shaham B, Wood BP, Bernstein BH, Stanley P, Szer IS.
12. Becker ML, van Haandel L, Gaedigk R, Lasky A, Hoeltzel M, High dose methotrexate in the treatment of refractory juvenile
Stobaugh J, et al. Analysis of intracellular methotrexate polyglu-
rheumatoid arthritis. Clin Exp Rheumatol 1995;13:113–8.
tamates in patients with juvenile idiopathic arthritis: effect of route
24. Stamp LK, O’Donnell JL, Chapman PT, Zhang M, Frampton C,
of administration on variability in intracellular methotrexate poly-
James J, et al. Determinants of red blood cell methotrexate
glutamate concentrations. Arthritis Rheum 2010;62:1803–12.
polyglutamate concentrations in rheumatoid arthritis patients re-
13. Petty RE, Southwood TR, Manners P, Baum J, Glass DN,
Goldenberg J, et al. International League of Associations for ceiving long-term methotrexate treatment. Arthritis Rheum 2009;
Rheumatology classification of juvenile idiopathic arthritis: second 60:2248–56.
revision, Edmonton, 2001. J Rheumatol 2004;31:390–2. 25. Dervieux T, Kremer J, Lein DO, Capps R, Barham R, Meyer G, et
14. Van Haandel L, Becker ML, Leeder JS, Williams TD, Stobaugh al. Contribution of common polymorphisms in reduced folate
JF. A novel high-performance liquid chromatography/mass spec- carrier and gamma-glutamylhydrolase to methotrexate polygluta-
trometry method for improved selective and sensitive measure- mate levels in patients with rheumatoid arthritis. Pharmacogenet-
ment of methotrexate polyglutamation status in human red blood ics 2004;14:733–9.
cells. Rapid Commun Mass Spectrom 2009;23:3693–702. 26. Stamp LK, O’Donnell JL, Chapman PT, Zhang M, James J,
15. Giannini EH, Ruperto N, Ravelli A, Lovell DJ, Felson DT, Frampton C, et al. Methotrexate polyglutamate concentrations are
Martini A. Preliminary definition of improvement in juvenile not associated with disease control in rheumatoid arthritis patients
arthritis. Arthritis Rheum 1997;40:1202–9. receiving long-term methotrexate therapy. Arthritis Rheum 2010;
16. Hahn LW, Ritchie MD, Moore JH. Multifactor dimensionality 62:359–68.
reduction software for detecting gene–gene and gene– 27. Hinks A, Moncrieffe H, Martin P, Lal SD, Ursu S, Kassoumeri L,
environment interactions. Bioinformatics 2003;19:376–82. et al. Genetic polymorphisms in key methotrexate (MTX) pathway
17. Giannini EH, Ruperto N, Ravelli A, Lovell DJ, Felson DT, genes associated with response to MTX treatment in juvenile
Martini A. Preliminary definition of improvement in juvenile idiopathic arthritis [abstract]. Arthritis Rheum 2009;60 Suppl:
arthritis. Arthritis Rheum 1997;40:1202–9. S233.
18. Ruperto N, Murray KJ, Gerloni V, Wulffraat N, de Oliveira SK, 28. Stocco G, Cheok MH, Crews KR, Dervieux T, French D, Pei D,
Falcini F, et al, for the Pediatric Rheumatology International et al. Genetic polymorphism of inosine triphosphate pyrophos-
Trials Organization. A randomized trial of parenteral methotrex- phatase is a determinant of mercaptopurine metabolism and
ate comparing an intermediate dose with a higher dose in children toxicity during treatment for acute lymphoblastic leukemia. Clin
with juvenile idiopathic arthritis who failed to respond to standard Pharmacol Ther 2009;85:164–72.
doses of methotrexate. Arthritis Rheum 2004;50:2191–201. 29. Dolezalova P, Krijt J, Chladek J, Nemcova D, Hoza J. Adenosine
19. Lambert CM, Sandhu S, Lochhead A, Hurst NP, McRorie E, and methotrexate polyglutamate concentrations in patients with
Dhillon V. Dose escalation of parenteral methotrexate in active juvenile arthritis. Rheumatology (Oxford) 2005;44:74–9.

You might also like